Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(1)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38203811

ABSTRACT

Extracellular vesicles (EVs) have a significant impact on the pathophysiological processes associated with various diseases such as tumors, inflammation, and infection. They exhibit molecular, biochemical, and entry control characteristics similar to viral infections. Viruses, on the other hand, depend on host metabolic machineries to fulfill their biosynthetic requirements. Due to potential advantages such as biocompatibility, biodegradation, and efficient immune activation, EVs have emerged as potential therapeutic targets against the SARS-CoV-2 infection. Studies on COVID-19 patients have shown that they frequently have dysregulated lipid profiles, which are associated with an increased risk of severe repercussions. Lipid droplets (LDs) serve as organelles with significant roles in lipid metabolism and energy homeostasis as well as having a wide range of functions in infections. The down-modulation of lipids, such as sphingolipid ceramide and eicosanoids, or of the transcriptional factors involved in lipogenesis seem to inhibit the viral multiplication, suggesting their involvement in the virus replication and pathogenesis as well as highlighting their potential as targets for drug development. Hence, this review focuses on the role of modulation of lipid metabolism and EVs in the mechanism of immune system evasion during SARS-CoV-2 infection and explores the therapeutic potential of EVs as well as application for delivering therapeutic substances to mitigate viral infections.


Subject(s)
COVID-19 , Extracellular Vesicles , Humans , Lipid Metabolism , SARS-CoV-2 , Lipogenesis
2.
Front Cell Infect Microbiol ; 13: 1102643, 2023.
Article in English | MEDLINE | ID: mdl-36909724

ABSTRACT

Tuberculosis (TB) is an infectious disease caused by the bacteria of the Mycobaterium tuberculosis (Mtb) complex. The modulation of the lipid metabolism has been implicated in the immune response regulation, including the formation of lipid droplets (LD)s, LD-phagosome association and eicosanoid synthesis. Mtb, M. bovis BCG and other pathogenic mycobacteria, as well as wall components, such as LAM, can induce LDs formation in a mechanism involving surface receptors, for instance TLRs, CD36, CD14, CD11b/CD18 and others. In addition, the activation of the lipid-activated nuclear receptor PPARγ is involved in the mechanisms of LD biogenesis, as well as in the modulation of the synthesis of lipid mediators. In infected cells, LDs are sites of compartmentalized prostaglandin E2 synthesis involved in macrophage deactivation, bacterial replication and regulation of the host cytokine profile. LDs also have a function in vesicle traffic during infection. Rab7 and RILP, but not Rab5, are located on LDs of infected macrophages, suggesting that LDs and phagosomes could exchange essential proteins for phagosomal maturation, interfering in mycobacterial survival. The pharmacological inhibition of LDs biogenesis affects the bacterial replication and the synthesis of lipid mediators and cytokines, suggesting that LDs may be new targets for antimicrobial therapies. However, it is still controversial if the accumulation of LDs favors the mycobacterial survival acting as an escape mechanism, or promotes the host resistance to infection. Thus, in this mini-review we discuss recent advances in understanding the important role of LDs in the course of infections and the implications for the pathophysiology of mycobacteriosis.


Subject(s)
Lipid Droplets , Tuberculosis , Humans , Lipid Droplets/metabolism , Tuberculosis/metabolism , Macrophages/microbiology , Phagosomes/metabolism , Lipid Metabolism , Lipids
3.
Front Cell Infect Microbiol ; 11: 768124, 2021.
Article in English | MEDLINE | ID: mdl-34778110

ABSTRACT

Chagas disease is a major public health problem, especially in the South and Central America region. Its incidence is related to poverty and presents a high rate of morbidity and mortality. The pathogenesis of Chagas disease is complex and involves many interactive pathways between the hosts and the Trypanosoma cruzi. Several factors have been implicated in parasite-host interactions, including molecules secreted by infected cells, lipid mediators and most recent, extracellular vesicles (EVs). The EVs of T. cruzi (EVsT) were reported for the first time in the epimastigote forms about 42 years ago. The EVsT are involved in paracrine communication during the infection and can have an important role in the inflammatory modulation and parasite escape mechanism. However, the mechanisms by which EVs employ their pathological effects are not yet understood. The EVsT seem to participate in the activation of macrophages via TLR2 triggering the production of cytokines and a range of other molecules, thus modulating the host immune response which promotes the parasite survival. Moreover, new insights have demonstrated that EVsT induce lipid body formation and PGE2 synthesis in macrophages. This phenomenon is followed by the inhibition of the synthesis of pro-inflammatory cytokines and antigen presentation, causing decreased parasitic molecules and allowing intracellular parasite survival. Therefore, this mini review aims to discuss the role of the EVs from T. cruzi as well as its involvement in the mechanisms that regulate the host immune response in the lipid metabolism and its significance for the Chagas disease pathophysiology.


Subject(s)
Chagas Disease , Extracellular Vesicles , Trypanosoma cruzi , Chagas Disease/metabolism , Extracellular Vesicles/metabolism , Humans , Immunity , Lipid Metabolism
4.
Spectrochim Acta A Mol Biomol Spectrosc ; 255: 119660, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33744843

ABSTRACT

Gold nanoparticles (AuNP) modified with antibody and rifampicin (RP) were tested against Mycobacterium bovis Bacillus Calmette-Guérin (BCG), which previously generated in vitro infection of macrophages from mice. Such a drug delivery system works as nanocarrier for RP and presented lower toxicity for macrophages cells than each separated component. Surface-enhanced Raman scattering (SERS) spectroscopy and fluorescence microscopy were used as analytical tools for the characterization of the internalization of gold nanocarriers into macrophage cells. The effective antibiotic action of RP, when combined with gold nanocarrier, was confirmed by dead-live assay of BCG bacteria lysed from macrophages after incubation. Such results indicate the delivery of RP to BCG bacteria, which were infecting macrophages, occurred with remarkable efficiency. It was rationalized based on the strategy used for the adsorption of antibody molecules on gold surface.


Subject(s)
Metal Nanoparticles , Mycobacterium bovis , Animals , Drug Delivery Systems , Gold , Macrophages , Mice , Spectrum Analysis, Raman
5.
Article in English | MEDLINE | ID: mdl-32229179

ABSTRACT

Lipid droplets (LDs) are organelles that have multiple roles in inflammatory and infectious diseases. LD act as essential platforms for immunometabolic regulation, including as sites for lipid storage and metabolism, inflammatory lipid mediator production, and signaling pathway compartmentalization. Accumulating evidence indicates that intracellular pathogens may exploit host LDs as source of nutrients and as part of their strategy to promote immune evasion. Notably, numerous studies have demonstrated the interaction between LDs and pathogen-containing phagosomes. However, the mechanism involved in this phenomenon remains elusive. Here, we observed LDs and PLIN2 surrounding M. bovis BCG-containing phagosomes, which included observations of a bacillus cell surrounded by lipid content inside a phagosome and LAM from mycobacteria co-localizing with LDs; these results were suggestive of exchange of contents between these compartments. By using beads coated with M.tb lipids, we demonstrated that LD-phagosome associations are regulated through the mycobacterial cell wall components LAM and PIM. In addition, we demonstrated that Rab7 and RILP, but not Rab5, localizes to LDs of infected macrophages and observed the presence of Rab7 at the site of interaction with an infected phagosome. Moreover, treatment of macrophages with the Rab7 inhibitor CID1067700 significantly inhibited the association between LDs and LAM-coated beads. Altogether, our data demonstrate that LD-phagosome interactions are controlled by mycobacterial cell wall components and Rab7, which enables the exchange of contents between LDs and phagosomes and may represent a fundamental aspect of bacterial pathogenesis and immune evasion.


Subject(s)
Lipid Droplets/metabolism , Mycobacterium Infections/metabolism , Mycobacterium tuberculosis/metabolism , Phagosomes/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/cytology , rab7 GTP-Binding Proteins
6.
J Eukaryot Microbiol ; 66(3): 385-392, 2019 05.
Article in English | MEDLINE | ID: mdl-30076737

ABSTRACT

Since the observation of the great pleomorphism of fish trypanosomes, in vitro culture has become an important tool to support taxonomic studies investigating the biology of cultured parasites, such as their structure, growth dynamics, and cellular cycle. Relative to their biology, ex vivo and in vitro studies have shown that these parasites, during the multiplication process, duplicate and segregate the kinetoplast before nucleus replication and division. However, the inverse sequence (the nucleus divides before the kinetoplast) has only been documented for a species of marine fish trypanosomes on a single occasion. Now, this previously rare event was observed in Trypanosoma abeli, a freshwater fish trypanosome. Specifically, from 376 cultured parasites in the multiplication process, we determined the sequence of organelle division for 111 forms; 39% exhibited nucleus duplication prior to kinetoplast replication. Thus, our results suggest that nucleus division before the kinetoplast may not represent an accidental or erroneous event occurring in the main pathway of parasite reproduction, but instead could be a species-specific process of cell biology in trypanosomes, such as previously noticed for Leishmania. This "alternative" pathway for organelle replication is a new field to be explored concerning the biology of marine and freshwater fish trypanosomes.


Subject(s)
Organelles/physiology , Trypanosoma/physiology , Animals , Catfishes/parasitology , Cell Division/physiology , Fish Diseases/parasitology , Trypanosomiasis/parasitology , Trypanosomiasis/veterinary
7.
J Eukaryot Microbiol, v. 66, n. 3, p. 385-392, mai/jun 2019
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2740

ABSTRACT

Since the observation of the great pleomorphism of fish trypanosomes, in vitroculture has become an important tool to support taxonomic studies investigat-ing the biology of cultured parasites, such as their structure, growth dynamics,and cellular cycle. Relative to their biology, ex vivo and in vitro studies haveshown that these parasites, during the multiplication process, duplicate andsegregate the kinetoplast before nucleus replication and division. However,the inverse sequence (the nucleus divides before the kinetoplast) has onlybeen documented for a species of marine fish trypanosomes on a single occa-sion. Now, this previously rare event was observed inTrypanosoma abeli,afreshwater fish trypanosome. Specifically, from 376 cultured parasites in themultiplication process, we determined the sequence of organelle division for111 forms; 39% exhibited nucleus duplication prior to kinetoplast replication.Thus, our results suggest that nucleus division before the kinetoplast may notrepresent an accidental or erroneous event occurring in the main pathway ofparasite reproduction, but instead could be a species-specific process of cellbiology in trypanosomes, such as previously noticed forLeishmania. This "al-ternative" pathway for organelle replication is a new field to be explored con-cerning the biology of marine and freshwater fish trypanosomes.

8.
J Eukaryot Microbiol ; 66(6): p. 385–392, 2019.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib15980

ABSTRACT

Since the observation of the great pleomorphism of fish trypanosomes, in vitroculture has become an important tool to support taxonomic studies investigat-ing the biology of cultured parasites, such as their structure, growth dynamics,and cellular cycle. Relative to their biology, ex vivo and in vitro studies haveshown that these parasites, during the multiplication process, duplicate andsegregate the kinetoplast before nucleus replication and division. However,the inverse sequence (the nucleus divides before the kinetoplast) has onlybeen documented for a species of marine fish trypanosomes on a single occa-sion. Now, this previously rare event was observed inTrypanosoma abeli,afreshwater fish trypanosome. Specifically, from 376 cultured parasites in themultiplication process, we determined the sequence of organelle division for111 forms; 39% exhibited nucleus duplication prior to kinetoplast replication.Thus, our results suggest that nucleus division before the kinetoplast may notrepresent an accidental or erroneous event occurring in the main pathway ofparasite reproduction, but instead could be a species-specific process of cellbiology in trypanosomes, such as previously noticed forLeishmania. This "al-ternative" pathway for organelle replication is a new field to be explored con-cerning the biology of marine and freshwater fish trypanosomes.

10.
Front Microbiol ; 9: 499, 2018.
Article in English | MEDLINE | ID: mdl-29616011

ABSTRACT

During Chagas disease, the Trypanosoma cruzi can induce some changes in the host cells in order to escape or manipulate the host immune response. The modulation of the lipid metabolism in the host phagocytes or in the parasite itself is one feature that has been observed. The goal of this mini review is to discuss the mechanisms that regulate intracellular lipid body (LB) biogenesis in the course of this parasite infection and their meaning to the pathophysiology of the disease. The interaction host-parasite induces LB (or lipid droplet) formation in a Toll-like receptor 2-dependent mechanism in macrophages and is enhanced by apoptotic cell uptake. Simultaneously, there is a lipid accumulation in the parasite due to the incorporation of host fatty acids. The increase in the LB accumulation during infection is correlated with an increase in the synthesis of PGE2 within the host cells and the parasite LBs. Moreover, the treatment with fatty acid synthase inhibitor C75 or non-steroidal anti-inflammatory drugs such as NS-398 and aspirin inhibited the LB biogenesis and also induced the down modulation of the eicosanoid production and the parasite replication. These findings show that LBs are organelles up modulated during the course of infection. Furthermore, the biogenesis of the LB is involved in the lipid mediator generation by both the macrophages and the parasite triggering escape mechanisms.

11.
Front Immunol ; 9: 111, 2018.
Article in English | MEDLINE | ID: mdl-29467755

ABSTRACT

Leptin directly activates macrophages and lymphocytes, but the role of leptin in neutrophil activation and migration is still controversial. Here, we investigate the in vivo mechanisms of neutrophil migration induced by leptin. The intraperitoneal injection of leptin (1 mg/kg) induces a time- and concentration-dependent neutrophil influx. We did not observe the enhancement of lipid bodies/droplets in neutrophils, after leptin treatment, as we had observed previously in peritoneal macrophages. The participation of leukotriene B4 (LTB4) in neutrophil recruitment triggered by leptin was investigated using different strategies. Leptin-induced neutrophil recruitment occurs both in the absence of 5-lipoxygenase activity in 5-lipoxygenase (5-LO)-/- mice and after the administration of either 5-LO inhibitor (Zileuton) or the LTB4 receptor antagonist (U-75302). Moreover, no direct induction of LTB4 by leptin could be observed. Neutrophil influx could not be prevented by the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, contrasting with the leptin-induced signaling for lipid body formation in macrophage that is mTOR-dependent. Leptin administration led to tumor necrosis factor-alpha (TNFα) production by the peritoneal cells both in vivo and in vitro. In addition, neutrophil recruitment was inhibited in tumor necrosis factor receptor 1 (TNFR1-/-) mice, indicating a role for TNF in leptin-induced neutrophil recruitment to the peritoneal cavity. Leptin-induced neutrophil influx was PI3Kγ-dependent, as it was absent in PI3Kγ-/- mice. Accordingly, leptin induced the peritoneal cells to produce CXCL1, both in vivo and in vitro, and the neutrophil influx was ablated after using an antibody against CXCL1. Our results establish TNFα/TNFR1- and CXCL1-dependent signaling as important pathways for leptin-induced neutrophil migration in vivo.


Subject(s)
Chemokine CXCL1/physiology , Leptin/physiology , Neutrophils/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Arachidonate 5-Lipoxygenase/genetics , Cell Movement , Chemokine CCL3/genetics , Macrophages, Peritoneal/immunology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Phosphatidylinositol 3-Kinases/genetics
12.
PLoS One ; 11(8): e0160433, 2016.
Article in English | MEDLINE | ID: mdl-27490663

ABSTRACT

Most eukaryotic cells contain varying amounts of cytosolic lipidic inclusions termed lipid bodies (LBs) or lipid droplets (LDs). In mammalian cells, such as macrophages, these lipid-rich organelles are formed in response to host-pathogen interaction during infectious diseases and are sites for biosynthesis of arachidonic acid (AA)-derived inflammatory mediators (eicosanoids). Less clear are the functions of LBs in pathogenic lower eukaryotes. In this study, we demonstrated that LBs, visualized by light microscopy with different probes and transmission electron microscopy (TEM), are produced in trypomastigote forms of the parasite Trypanosoma cruzi, the causal agent of Chagas' disease, after both host interaction and exogenous AA stimulation. Quantitative TEM revealed that LBs from amastigotes, the intracellular forms of the parasite, growing in vivo have increased size and electron-density compared to LBs from amastigotes living in vitro. AA-stimulated trypomastigotes released high amounts of prostaglandin E2 (PGE2) and showed PGE2 synthase expression. Raman spectroscopy demonstrated increased unsaturated lipid content and AA incorporation in stimulated parasites. Moreover, both Raman and MALDI mass spectroscopy revealed increased AA content in LBs purified from AA-stimulated parasites compared to LBs from unstimulated group. By using a specific technique for eicosanoid detection, we immunolocalized PGE2 within LBs from AA-stimulated trypomastigotes. Altogether, our findings demonstrate that LBs from the parasite Trypanosoma cruzi are not just lipid storage inclusions but dynamic organelles, able to respond to host interaction and inflammatory events and involved in the AA metabolism. Acting as sources of PGE2, a potent immunomodulatory lipid mediator that inhibits many aspects of innate and adaptive immunity, newly-formed parasite LBs may be implicated with the pathogen survival in its host.


Subject(s)
Arachidonic Acid/metabolism , Lipid Droplets/metabolism , Trypanosoma cruzi/metabolism , Chagas Disease/metabolism , Dinoprostone/metabolism , Lipid Droplets/ultrastructure , Prostaglandin-E Synthases/biosynthesis , Protozoan Proteins/biosynthesis , Trypanosoma cruzi/ultrastructure
13.
Front Immunol ; 7: 174, 2016.
Article in English | MEDLINE | ID: mdl-27199996

ABSTRACT

Pathogens induce several changes in the host cell signaling and trafficking mechanisms in order to evade and manipulate the immune response. One prominent pathogen-mediated change is the formation of lipid-rich organelles, termed lipid bodies (LBs) or lipid droplets, in the host cell cytoplasm. Protozoan parasites, which contribute expressively to the burden of infectious diseases worldwide, are able to induce LB genesis in non-immune and immune cells, mainly macrophages, key players in the initial resistance to the infection. Under host-parasite interaction, LBs not only accumulate in the host cytoplasm but also relocate around and move into parasitophorous vacuoles. There is increasing evidence that protozoan parasites may target host-derived LBs either for gaining nutrients or for escaping the host immune response. Newly formed, parasite-induced LBs may serve as lipid sources for parasite growth and also produce inflammatory mediators that potentially act in the host immune response deactivation. In this mini review, we summarize current knowledge on the formation and role of host LBs as sites exploited by intracellular protozoan parasites as a strategy to maintain their own survival.

14.
Mem Inst Oswaldo Cruz ; 109(6): 767-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25317704

ABSTRACT

Lipid bodies [lipid droplets (LBs)] are lipid-rich organelles involved in lipid metabolism, signalling and inflammation. Recent findings suggest a role for LBs in host response to infection; however, the potential functions of this organelle in Toxoplasma gondii infection and how it alters macrophage microbicidal capacity during infection are not well understood. Here, we investigated the role of host LBs in T. gondii infection in mouse peritoneal macrophages in vitro. Macrophages cultured with mouse serum (MS) had higher numbers of LBs than those cultured in foetal bovine serum and can function as a model to study the role of LBs during intracellular pathogen infection. LBs were found in association with the parasitophorous vacuole, suggesting that T. gondii may benefit from this lipid source. Moreover, increased numbers of macrophage LBs correlated with high prostaglandin E2 (PGE2) production and decreased nitric oxide (NO) synthesis. Accordingly, LB-enriched macrophages cultured with MS were less efficient at controlling T. gondii growth. Treatment of macrophages cultured with MS with indomethacin, an inhibitor of PGE2 production, increased the microbicidal capacity against T. gondii. Collectively, these results suggest that culture with MS caused a decrease in microbicidal activity of macrophages against T. gondii by increasing PGE2 while lowering NO production.


Subject(s)
Lipid Droplets/parasitology , Macrophage Activation/physiology , Macrophages, Peritoneal/parasitology , Toxoplasma/physiology , Vacuoles/parasitology , Animals , Cattle , Host-Parasite Interactions , Indomethacin/pharmacology , Lipid Droplets/physiology , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/physiology , Macrophages, Peritoneal/ultrastructure , Male , Mice , Mice, Inbred C3H , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nitric Oxide/biosynthesis , Primary Cell Culture , Prostaglandins E/antagonists & inhibitors , Prostaglandins E/biosynthesis , Vacuoles/physiology
15.
Mediators Inflamm ; 2012: 956509, 2012.
Article in English | MEDLINE | ID: mdl-23209347

ABSTRACT

Oleic acid (OA) can induce acute lung injury in experimental models. In the present work, we used intratracheal OA injection to show augmented oedema formation, cell migration and activation, lipid mediator, and cytokine productions in the bronchoalveolar fluids of Swiss Webster mice. We also demonstrated that OA-induced pulmonary injury is dependent on ERK1/2 activation, since U0126, an inhibitor of ERK1/2 phosphorylation, blocked neutrophil migration, oedema, and lipid body formation as well as IL-6, but not IL-1ß production. Using a mice strain carrying a null mutation for the TLR4 receptor, we proved that increased inflammatory parameters after OA challenges were not due to the activation of the TLR4 receptor. With OA being a Na/K-ATPase inhibitor, we suggest the possible involvement of this enzyme as an OA target triggering lung inflammation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Injury/chemically induced , MAP Kinase Signaling System/drug effects , Oleic Acid/toxicity , Animals , Cytokines/physiology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Phosphorylation , Pulmonary Edema/etiology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Toll-Like Receptor 4/physiology
16.
Mediators Inflamm ; 2012: 478601, 2012.
Article in English | MEDLINE | ID: mdl-22619483

ABSTRACT

The flagellated protozoa Trypanosoma cruzi is the causal agent of Chagas' disease, a significant public health issue and still a major cause of morbidity and mortality in Latin America. Acute Chagas' disease elicits a strong inflammatory response. In order to control the parasite multiplication, cells of the monocytic lineage are highly mobilized. Monocyte differentiation leads to the formation of phagocytosing macrophages, which are strongly activated and direct host defense. A distinguishing feature of Chagas' disease-triggered macrophages is the presence of increased numbers of distinct cytoplasmic organelles termed lipid bodies or lipid droplets. These organelles are actively formed in response to the parasite and are sites for synthesis and storage of inflammatory mediators. This review covers current knowledge on lipid bodies elicited by the acute Chagas' disease within inflammatory macrophages and discusses the role of these organelles in inflammation. The increased knowledge of lipid bodies in pathogenic mechanisms of infections may not only contribute to the understanding of pathogen-host interactions but may also identify new targets for intervention.


Subject(s)
Chagas Disease/parasitology , Host-Pathogen Interactions , Monocytes/parasitology , Trypanosoma cruzi/physiology , Animals , Apoptosis , Chagas Disease/immunology , Coculture Techniques , Cytoplasm/metabolism , Disease Models, Animal , Humans , Immunity, Innate , Inflammation , Lipids/chemistry , Macrophages/immunology , Macrophages/parasitology , Mice , Microscopy, Fluorescence/methods , Monocytes/cytology , Organelles/parasitology , Rats , Trypanosoma cruzi/metabolism
17.
J Immunol ; 187(5): 2548-58, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21813774

ABSTRACT

The mechanisms responsible for nerve injury in leprosy need further elucidation. We recently demonstrated that the foamy phenotype of Mycobacterium leprae-infected Schwann cells (SCs) observed in nerves of multibacillary patients results from the capacity of M. leprae to induce and recruit lipid droplets (LDs; also known as lipid bodies) to bacterial-containing phagosomes. In this study, we analyzed the parameters that govern LD biogenesis by M. leprae in SCs and how this contributes to the innate immune response elicited by M. leprae. Our observations indicated that LD formation requires the uptake of live bacteria and depends on host cell cytoskeleton rearrangement and vesicular trafficking. TLR6 deletion, but not TLR2, completely abolished the induction of LDs by M. leprae, as well as inhibited the bacterial uptake in SCs. M. leprae-induced LD biogenesis correlated with increased PGE(2) and IL-10 secretion, as well as reduced IL-12 and NO production in M. leprae-infected SCs. Analysis of nerves from lepromatous leprosy patients showed colocalization of M. leprae, LDs, and cyclooxygenase-2 in SCs, indicating that LDs are sites for PGE(2) synthesis in vivo. LD biogenesis Inhibition by the fatty acid synthase inhibitor C-75 abolished the effect of M. leprae on SC production of immunoinflammatory mediators and enhanced the mycobacterial-killing ability of SCs. Altogether, our data indicated a critical role for TLR6-dependent signaling in M. leprae-SC interactions, favoring phagocytosis and subsequent signaling for induction of LD biogenesis in infected cells. Moreover, our observations reinforced the role of LDs favoring mycobacterial survival and persistence in the nerve. These findings give further support to a critical role for LDs in M. leprae pathogenesis in the nerve.


Subject(s)
Leprosy/pathology , Schwann Cells/microbiology , Schwann Cells/pathology , Toll-Like Receptor 6/immunology , Animals , Humans , Immunohistochemistry , Inclusion Bodies/immunology , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Leprosy/immunology , Lipid Metabolism/physiology , Lipids/immunology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Mycobacterium leprae/immunology , Schwann Cells/immunology , Toll-Like Receptor 6/metabolism
18.
J Infect Dis ; 204(6): 951-61, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21849292

ABSTRACT

Lipid bodies (lipid droplets) are lipid-rich organelles with functions in cell metabolism and signaling. Here, we investigate the mechanisms of Trypanosoma cruzi-induced lipid body formation and their contributions to host-parasite interplay. We demonstrate that T. cruzi-induced lipid body formation in macrophages occurs in a Toll-like receptor 2-dependent mechanism and is potentiated by apoptotic cell uptake. Lipid body biogenesis and prostaglandin E2 (PGE2) production triggered by apoptotic cell uptake was largely dependent of α(v)ß3 and transforming growth factor-ß signaling. T. cruzi-induced lipid bodies act as sites of increased PGE synthesis. Inhibition of lipid body biogenesis by the fatty acid synthase inhibitor C75 reversed the effects of apoptotic cells on lipid body formation, eicosanoid synthesis, and parasite replication. Our findings indicate that lipid bodies are highly regulated organelles during T. cruzi infection with roles in lipid mediator generation by macrophages and are potentially involved in T. cruzi-triggered escape mechanisms.


Subject(s)
Chagas Disease/pathology , Dinoprostone/metabolism , Host-Parasite Interactions , Lipid Metabolism , Macrophages/metabolism , Macrophages/parasitology , Trypanosoma cruzi/pathogenicity , Animals , Female , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 2/metabolism , Trypanosoma cruzi/growth & development
19.
J Histochem Cytochem ; 59(5): 540-56, 2011 May.
Article in English | MEDLINE | ID: mdl-21430261

ABSTRACT

Lipid bodies (LBs), also known as lipid droplets, have increasingly been recognized as functionally active organelles linked to diverse biological functions and human diseases. These organelles are actively formed in vivo within cells from the immune system, such as macrophages, neutrophils, and eosinophils, in response to different inflammatory conditions and are sites for synthesis and storage of inflammatory mediators. In this review, the authors discuss structural and functional aspects of LBs and current imaging techniques to visualize these organelles in cells engaged in inflammatory processes, including infectious diseases. The dynamic morphological aspects of LBs in leukocytes as inducible, newly formable organelles, elicitable in response to stimuli that lead to cellular activation, contribute to the evolving understanding of LBs as organelles that are critical regulators of different inflammatory diseases, key markers of leukocyte activation, and attractive targets for novel anti-inflammatory therapies.


Subject(s)
Inflammation/metabolism , Lipid Metabolism , Organelles/physiology , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Chagas Disease/immunology , Chagas Disease/metabolism , Hepatitis C/immunology , Hepatitis C/metabolism , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Leukocytes/metabolism , Macrophages/metabolism , Microscopy/methods , Organelles/ultrastructure , Spectrum Analysis, Raman , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism
20.
Cell Microbiol ; 13(2): 259-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20955239

ABSTRACT

The predilection of Mycobacterium leprae (ML) for Schwann cells (SCs) leads to peripheral neuropathy, a major concern in leprosy. Highly infected SCs in lepromatous leprosy nerves show a foamy, lipid-laden appearance; but the origin and nature of these lipids, as well as their role in leprosy, have remained unclear. The data presented show that ML has a pronounced effect on host-cell lipid homeostasis through regulation of lipid droplet (lipid bodies, LD) biogenesis and intracellular distribution. Electron microscopy and immunohistochemical analysis of lepromatous leprosy nerves for adipose differentiation-related protein expression, a classical LD marker, revealed accumulating LDs in close association to ML in infected SCs. The capacity of ML to induce LD formation was confirmed in in vitro studies with human SCs. Moreover, via confocal and live-cell analysis, it was found that LDs are promptly recruited to bacterial phagosomes and that this process depends on cytoskeletal reorganization and PI3K signalling. ML-induced LD biogenesis and recruitment were found to be independent of TLR2 bacterial sensing. Notably, LD recruitment impairment by cytoskeleton drugs decreased intracellular bacterial survival. Altogether, our data revealed SC lipid accumulation in ML-containing phagosomes, which may represent a fundamental aspect of bacterial pathogenesis in the nerve.


Subject(s)
Lipid Metabolism , Mycobacterium leprae/pathogenicity , Phagosomes/microbiology , Schwann Cells/microbiology , Cells, Cultured , Cytoplasm/chemistry , Cytoplasm/ultrastructure , Cytoskeleton/metabolism , Humans , Immunohistochemistry , Membrane Proteins/analysis , Microbial Viability , Microscopy , Mycobacterium leprae/metabolism , Perilipin-2 , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...